P53 Signaling Modulation of Cell Cycle Arrest and Viral Replication In

Total Page:16

File Type:pdf, Size:1020Kb

P53 Signaling Modulation of Cell Cycle Arrest and Viral Replication In Xu et al. Vet Res (2016) 47:120 DOI 10.1186/s13567-016-0403-4 RESEARCH ARTICLE Open Access p53 signaling modulation of cell cycle arrest and viral replication in porcine circovirus type 2 infection cells Dan Xu†, Qian Du†, Cong Han, Zengguo Wang, Xiujuan Zhang, Tongtong Wang, Xiaomin Zhao, Yong Huang* and Dewen Tong* Abstract Porcine circovirus type 2 (PCV2) is a ubiquitous pathogen in the swine industry worldwide. Previous studies have shown that PCV2 infection induces host cell apoptosis through up-regulation of p53. To further identify the regula- tory roles of p53 signaling in the process of PCV2 infection, we established p53 gene knockout PK15 cell lines using the genomic editor tool CRISPR/Cas9, and further investigated the roles of p53 in modulating the cell cycle and viral replication in this study. The results show that PCV2 infection induced obvious S phase accumulation in wild-type PK15 cells and a compromised S phase accumulation in the p53 gene mutation cells (813PK15p53m/m), but did not p53 / induce obvious S phase accumulation in the p53 gene knockout cells (148PK15 − −) compared with the respec- tive mock infection. PCV2 infection activated p53 signaling, up-regulated the expression of p21, Cyclin E, and down- regulated Cyclin A, CDK2. In p53 deficient cells, however, PCV2-induced changes in Cyclin A, CDK2, and Cyclin E were efficiently reversed to the basal levels. Detection of PCV2 replication showed decreased viral ORF1 genomic DNA in p533 / p53m/m p53 deficient cells (148PK15 − −) and p53 mutated cells (813PK15 ) compared with p53 wild-type cells after different synchronization treatment. Furthermore, PCV2 viral genomic DNA and Cap protein levels were higher in the cells released from S phase synchronized cells than in the cells released from the G0/G1 phase or G2/M phase-syn- chronized, or asynchronous cells after 18 h post-infection. Taken together, this study demonstrates that PCV2 infection induces S phase accumulation to favor viral replication in host cells through activation of the p53 pathway. Introduction However, the roles of p53 signaling in modulating cell PCV2, belonging to the family Circoviridae, is the main cycle and PCV2 replication has not been defined up to pathogen to cause porcine circovirus associated diseases date. (PCVAD) [1], posing a huge threat for world pig hus- Numbers of studies have broadened our understand- bandry [2]. As a tiny DNA virus, PCV2 infection requires ing of the roles of p53 signaling in the process of different host cells to provide necessary resources for replication virus infection and replication. For instance, Kaposi’s sar- themselves, thus disturbing a variety of cell signaling coma herpesvirus (KSHV) activates host p53 signal and pathways to modulate the host cell cycle, proliferation, induces G2 phase arrest to promote the onset of virus survival and death to facilitate their infection and repli- replication [6]. Prototype foamy virus (PFV) promotes cation [3, 4]. Among the signaling pathways, p53 signal- p53 level increase by knockdown of Pirh2 to contrib- ing is essential for control of quiescent cell entry into the ute to the latency of PFV infection [7]. Herpes simplex cell cycle, and regulating cellular DNA replication [5]. virus type 2 infection can phosphorylate p53 protein to induce the G0/G1 phase arrest [8]. PRRSV manipulates the host factors mdm2 and p53 via its Nsp1α to increase *Correspondence: [email protected]; [email protected] viral replication at the early stage of infection [9]. Indeed, † Dan Xu and Qian Du contributed equally to this work previous studies have shown that PCV2 ORF3 protein College of Veterinary Medicine, Northwest A&F University, 22 Xinong Rd, Yangling, Shaanxi 712100, People’s Republic of China specifically interacts with porcine ubiquitin E3 ligase © The Author(s) 2016. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/ publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Xu et al. Vet Res (2016) 47:120 Page 2 of 11 Pirh2 to promote p53 accumulation [10], playing an CRISPR/cas9 KO cell important role in PCV2 pathogenesis [11], which indi- Targeting sites in the p53 gene were selected using the cates the key role of p53 in the interaction of PCV2 and CRISPR program (Genome Engineering. Broad Institute the host. However, in-depth study of the roles of p53 Cambridge, MA, USA) Oligonucleotide pairs for the target signaling in the process of PCV2 was limited due to lack- sequences were annealed and the resulting fragments were ing of p53 deficient cell line in porcines. then cloned into the BsmB I sites of lentiCRISPRv2 plasmid In this study, with the help of the CRISPR/Cas9 system, (Addgene), and co-transfected into HEK293T cells with the we constructed p53 deficient and mutant porcine cell packaging plasmids psPAX2 (AddGene 12260) to generate lines, and further detected and compared the difference the lentivirus. 72 h after the transfection, the supernatant of cell cycle profiles and viral replication between the was collected after three cycles of frozen-thawed. Titers of p53 wild-type, p53 deficient and p53 mutant porcine cell the obtained lentivirus expressing the target sequences were lines. This study allows us to deeply explore and confirm determined by qPCR. Finally, the CRISPR/Cas9 mediated the roles of p53 signaling in modulating cell cycle and P53 knockout cells were selected from lentivirus infected PCV2 replication. PK15 cell lines that were cultured in puromycin (500 ng/ mL) DMEM medium for at least 14 days. Genomic DNA Materials and methods sequence from PK15 cells was determined using primers: Cells, virus and antibodies 148-F: 5′-GACTCCTGTTGTTCCCATCCA-3′; 148-R: Porcine kidney 15 (PK15) cells purchased from ATCC 5′-AGGGAGCCAGCAGTCAAATG-3′; 813-F: 5′-GGGA (CCL-33) were cultured in Dulbecco’s Modified Eagle’s CGGAACAGCTTTGAGGT-3′; 813-R: 5′-CTGTTGGCA Medium (Gibco BRL, Gaithersburg, MD, USA) supple- AATGCCCCAAA-3′. mented with 10% heat-inactivated fetal bovine serum (Thermo Scientific HyClone, Beijing, China), and incu- Cell synchronization bated at 37 °C in a 5% CO2 atmosphere incubator. The Cells synchronized in G1/G0 phase were obtained by PCV2 strains (GenBank No. EU366323) used in this serum starvation. PK-15 cells were cultured in serum- study were isolated and purified previously by our team free medium for 24 h or 48 h, and then cells were washed and stocked in our laboratory, the UV-inactivation was with PBS and plated in fresh media to start PCV2 incuba- performed by UV radiation of the virus for 45 min in the tion for 1 h and cultured in 2% FBS DMEM medium for hood. The anti-PCV2 Cap primary antibodies were pro- 18 or 24 h for later analysis. Double thymidine block was duced by our team [12, 13]. The primary monoclonal rab- used for early S phase synchronization. The cells were bit antibodies of p53, p21 and anti-BrdU were purchased treated for 12 h with 2 mM thymidine, after which cells from Cell Signaling (Cell Signaling Technology, Danvers, were washed and released into fresh media with MOI = 1 MA, USA). CDK2, Cyclin A and Cyclin E antibodies PCV2 virus then incubated for 1 h, and cultured in 2% were purchased from Santa Cruz Biotechnology (Santa FBS DMEM medium for 18 h. The cells were treated with Cruz, California, CA, USA). The monoclonal antibody 100 ng/mL nocodazole for 16 h until arrest at the G2/M of β-actin was purchased from sigma (Sigma-Aldrich, phase, then the cells were released by washing with PBS St. Louis, MO, USA). The FITC goat anti-mouse IgG was and plated in fresh media to start PCV2 incubation for purchased from BD Biosciences (BD, San Jose, CA, USA). 1 h and culture in 2% FBS DMEM medium for 18 h for later analysis. Cell cycle analysis The ratio of cells in each phase of the cell cycle was deter- Detection of virus replication mined by DNA content using propidium iodide (PI) The cells were seeded in culture plates at a density of staining followed by flow cytometric analysis. The cells 1 106 cells/well, and cultured to reach approximately 6 × plated at a density of 1 × 10 cells/flask were treated 60–70% confluence. PCV2 strains were used to infect the with the indicated Multiplicity of infection (MOI) of cells at a multiplicity of infection of 1. Viral ORF1 frag- PCV2 for the indicated times as described in the figure ments were determined in each of the PK15 cell lines legends. The cells were trypsinized, washed twice with using primer PCV-F: 5′-AGTACCGGGAGTGGTAGG ′ ′ ′ PBS, and fixed with 70% ice-cold ethanol at− 20 °C over- AG-3 ; PCV-R: 5 -GTTGAATTCTGGCCCTGCTC-3 . night. Fixed cells were washed with cold PBS and resus- The supernatant and the attached cells were collected pended with PI staining solution containing 50 mg/mL together to extract the DNA. PI (Sigma-Aldrich), 100 mg/mL RNase A (TIANGEN Biotech, Beijing, China), and incubated in the dark for BrdU incorporation assay 30 min. The samples were analyzed using a flow cytom- For labeling of S-phase cells, BrdU was added in mid- eter (Accuri™ C6, BD Biosciences, San Diego, CA, USA). log phase cells at a final concentration of 10 µM and Xu et al. Vet Res (2016) 47:120 Page 3 of 11 incubated for 1 h at 37 °C.
Recommended publications
  • Discriminating the Eight Genotypes of the Porcine Circovirus Type 2 With
    Link et al. Virol J (2021) 18:70 https://doi.org/10.1186/s12985-021-01541-z RESEARCH Open Access Discriminating the eight genotypes of the porcine circovirus type 2 with TaqMan-based real-time PCR Ellen Kathrin Link1, Matthias Eddicks2, Liangliang Nan1, Mathias Ritzmann2, Gerd Sutter1 and Robert Fux1* Abstract Background: The porcine circovirus type 2 (PCV2) is divided into eight genotypes including the previously described genotypes PCV2a to PCV2f and the two new genotypes PCV2g and PCV2h. PCV2 genotyping has become an impor- tant task in molecular epidemiology and to advance research on the prophylaxis and pathogenesis of PCV2 associ- ated diseases. Standard genotyping of PCV2 is based on the sequencing of the viral genome or at least of the open reading frame 2. Although, the circovirus genome is small, classical sequencing is time consuming, expensive, less sensitive and less compatible with mass testing compared with modern real-time PCR assays. Here we report about a new PCV2 genotyping method using qPCR. Methods: Based on the analysis of several hundred PCV2 full genome sequences, we identifed PCV2 genotype specifc sequences or single-nucleotide polymorphisms. We designed six TaqMan PCR assays that are specifc for single genotypes PCV2a to PCV2f and two qPCRs targeting two genotypes simultaneously (PCV2g/PCV2d and PCV2h/ PCV2c). To improve specifc binding of oligonucleotide primers and TaqMan probes, we used locked nucleic acid technology. We evaluated amplifcation efciency, diagnostic sensitivity and tested assay specifcity for the respective genotypes. Results: All eight PCV2 genotype specifc qPCRs demonstrated appropriate amplifcation efciencies between 91 and 97%. Testing samples from an epidemiological feld study demonstrated a diagnostic sensitivity of the respective genotype specifc qPCR that was comparable to a highly sensitive pan-PCV2 qPCR system.
    [Show full text]
  • Epithelial Cell Death Analysis of Cell Cycle by Flow Cytometry White Paper
    Epithelial Cell Death Analysis of Cell Cycle by Flow Cytometry White Paper Authors: Savithri Balasubramanian, John Tigges, Vasilis Toxavidis, Heidi Mariani. Affiliation: Beth Israel Deaconess Medical Center Harvard Stem Cell Institute a Beckman Coulter Life Sciences: Epithelial Cell Death Analysis of Cell Cycle by Flow Cytometry PRINCIPAL OF TECHNIQUE Background: Cell cycle, or cell-division cycle, is the series of events that takes place in a cell leading to its division and duplication (replication). In cells without a nucleus (prokaryotic), cell cycle occurs via a process termed binary fission. In cells with a nucleus (eukaryotes), cell cycle can be divided in two brief periods: interphase—during which the cell grows, accumulating nutrients needed for mitosis and duplicating its DNA—and the mitosis (M) phase, during which the cell splits itself into two distinct cells, often called «daughter cells». Cell-division cycle is a vital process by which a single-celled fertilized egg develops into a mature organism, as well as the process by which hair, skin, blood cells, and some internal organs are renewed. Cell cycle consists of four distinct phases: G1 phase, S phase (synthesis), G2 phase (collectively known as interphase) and M phase (mitosis). M phase is itself composed of two tightly coupled processes: mitosis, in which the cell’s chromosomes are divided between the two daughter cells, and cytokinesis, in which the cell’s cytoplasm divides in half forming distinct cells. Activation of each phase is dependent on the proper progression and completion of the previous one. Cells that have temporarily or reversibly stopped dividing are said to have entered a state of quiescence called G0 phase.
    [Show full text]
  • Viral Diversity in Tree Species
    Universidade de Brasília Instituto de Ciências Biológicas Departamento de Fitopatologia Programa de Pós-Graduação em Biologia Microbiana Doctoral Thesis Viral diversity in tree species FLÁVIA MILENE BARROS NERY Brasília - DF, 2020 FLÁVIA MILENE BARROS NERY Viral diversity in tree species Thesis presented to the University of Brasília as a partial requirement for obtaining the title of Doctor in Microbiology by the Post - Graduate Program in Microbiology. Advisor Dra. Rita de Cássia Pereira Carvalho Co-advisor Dr. Fernando Lucas Melo BRASÍLIA, DF - BRAZIL FICHA CATALOGRÁFICA NERY, F.M.B Viral diversity in tree species Flávia Milene Barros Nery Brasília, 2025 Pages number: 126 Doctoral Thesis - Programa de Pós-Graduação em Biologia Microbiana, Universidade de Brasília, DF. I - Virus, tree species, metagenomics, High-throughput sequencing II - Universidade de Brasília, PPBM/ IB III - Viral diversity in tree species A minha mãe Ruth Ao meu noivo Neil Dedico Agradecimentos A Deus, gratidão por tudo e por ter me dado uma família e amigos que me amam e me apoiam em todas as minhas escolhas. Minha mãe Ruth e meu noivo Neil por todo o apoio e cuidado durante os momentos mais difíceis que enfrentei durante minha jornada. Aos meus irmãos André, Diego e meu sobrinho Bruno Kawai, gratidão. Aos meus amigos de longa data Rafaelle, Evanessa, Chênia, Tati, Leo, Suzi, Camilets, Ricardito, Jorgito e Diego, saudade da nossa amizade e dos bons tempos. Amo vocês com todo o meu coração! Minha orientadora e grande amiga Profa Rita de Cássia Pereira Carvalho, a quem escolhi e fui escolhida para amar e fazer parte da família.
    [Show full text]
  • Recruitment of Leukemic Cells from G 0 Phase of the Cell Cycle By
    Leukemia (2003) 17, 2049–2059 & 2003 Nature Publishing Group All rights reserved 0887-6924/03 $25.00 www.nature.com/leu CORRESPONDENCE Recruitment of leukemic cells from G0 phase of the cell cycle by interferons results in conversion of resistance to daunorubicin Leukemia (2003) 17, 2049–2051. doi:10.1038/sj.leu.2403085 were not affected by daunorubicin-induced cell death. To analyze whether a similar cell cycle-specific sensitivity to Ara-C TO THE EDITOR and daunorubicin was observed in other leukemic cell lines, we analyzed four acute lymphoblastic leukemia cell lines and one Although the majority of patients with acute myeloid leukemia CML blast crisis cell line, which were generated in our (AML) responds to initial treatment, relapse of the disease occurs laboratory by culturing of leukemic blasts from five different in a significant percentage of these patients.1 The cell cycle patients in serum-free medium at high cell concentrations until status of leukemic cells may play an important role in the spontaneous sustained proliferation of the leukemic cells response to treatment of leukemic cells. Especially, the broadly occurred. These cell lines cytogenetically and phenotypically used cytotoxic agent Cytarabine (Ara-C) has been demonstrated resembled the primary leukemia. Figure 1b shows the median to exert its action via intercalation into the DNA of cells cell cycle distribution within these cell lines after incubation for À6 specifically in the S phase of the cell cycle, and sensitivity to this 48 h in medium alone, or in medium containing 1 Â 10 M Ara- agent is therefore described to be specific for cells in active cell C or daunorubicin.
    [Show full text]
  • P53 Phosphorylation and Association with Murine Double Minute 2, C-Jun
    [CANCER RESEARCH 60, 896–900, February 15, 2000] Advances in Brief p53 Phosphorylation and Association with Murine Double Minute 2, c-Jun ARF NH2-Terminal Kinase, p14 , and p300/CBP during the Cell Cycle and after Exposure to Ultraviolet Irradiation1 Thomas Buschmann, Victor Adler, Ekaterina Matusevich, Serge Y. Fuchs, and Ze’ev Ronai2 Ruttenberg Cancer Center, Mount Sinai School of Medicine, New York, New York 10029 Abstract though the mechanisms underlying the ability of p53 to elicit such opposing effects are yet to be identified, independent studies point to p53 phosphorylation and association with proteins is implicated in its a different set of p53 regulators and effectors that are affected by p53 stability and activity. We have compared the association of DNA-bound in each of these scenarios. and overall pools of p53 with murine double minute 2 (Mdm2), c-Jun NH -terminal kinase (JNK), p300/CBP, and p14ARF during cell cycle In studying the regulation of p53 stability, we demonstrated previ- 2 ously that, in Swiss 3T3 cells, JNK and Mdm2 target p53 degradation progression. Whereas DNA-bound p53 associates with JNK at G0-G1 and in different phases of the cell cycle (7). In this study, we have with Mdm2 and p300 during S and G2-M phases, the general pool of p53 was found in complex with JNK and Mdm2 almost throughout the cell compared the association of subpopulations of p53 with proteins cycle. Phosphorylation of p53 at serines 9, 15, and 20 is at the highest levels implicated in stability and activity of p53 and monitored the pattern of at G1 and at serines 37 and 392 during G2-M phase.
    [Show full text]
  • Detection of Porcine Circoviruses in Clinical Specimens Using Multiplex PCR in Hubei, Central China
    Detection of porcine circoviruses in clinical specimens using multiplex PCR in Hubei, central China Keli Yang Corresp., 1 , Zuwu Jiao 1 , Danna Zhou 1 , Rui Guo 1 , Zhengying Duan 1 , Fangyan Yuan 1 , Yongxiang Tian 1 1 Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, China Corresponding Author: Keli Yang Email address: [email protected] In order to detect and simultaneously discriminate PCV1, PCV2 and PCV3, a multiplex PCR assay was developed and used to detect clinical samples in this study. Each of target genes of PCV1, PCV2 and PCV3 was amplified using the designed primers, while no other porcine viruses genes were detected. The limit of detection of the assay was 10 copies/μL of PCV1, PCV2 and PCV3. The tissue samples from eight pig farms were detected using the multiplex PCR assay. The results showed that PCV1, PCV2 and PCV3 are co-circulating in central China. The PCV1, PCV2 and PCV3 singular infection rate was 52.4% (150/286), 61.2% (175/286) and 45.1% (129/286), respectively, while the PCV1 and PCV2 co-infection rate was 11.2% (32/286), the PCV1 and PCV3 co-infection rate was 5.9% (17/286), the PCV2 and PCV3 co-infection rate was 23.4% (67/286), and the PCV1, PCV2 and PCV3 co- infection rate was 1.7% (5/286), respectively, which were 100% consistent with the sequencing method and Real-time PCR methods. It proved that this multiplex PCR assay could be used as a differential diagnostic tool for monitoring and control of PCVs in the field.
    [Show full text]
  • A P53-Responsive Mirna Network Promotes Cancer Cell Quiescence Ting La1, Guang Zhi Liu2, Margaret Farrelly1, Nicole Cole3, Yu Chen Feng1, Yuan Yuan Zhang1, Simonne K
    Published OnlineFirst October 9, 2018; DOI: 10.1158/0008-5472.CAN-18-1886 Cancer Tumor Biology and Immunology Research A p53-Responsive miRNA Network Promotes Cancer Cell Quiescence Ting La1, Guang Zhi Liu2, Margaret Farrelly1, Nicole Cole3, Yu Chen Feng1, Yuan Yuan Zhang1, Simonne K. Sherwin1, Hamed Yari1, Hessam Tabatabaee1, Xu Guang Yan1, Su Tang Guo4, Tao Liu5, Rick F. Thorne2,6, Lei Jin7, and Xu Dong Zhang1,2 Abstract Cancer cells in quiescence (G0 phase) are resistant to miRNA-455-3p targeted CDK2-associated cullin domain 1 death, and re-entry of quiescent cancer cells into the cell- (CAC1), which enhanced CDK2-mediated phosphorylation cycle plays an important role in cancer recurrence. Here we of p27 necessary for its polyubiquitination. Of note, the show that two p53-responsive miRNAs utilize distinct but gene encoding miRNA-27b-3p was embedded in the intron complementary mechanisms to promote cancer cell quies- of the chromosome 9 open reading frame 3 gene that was cence by facilitating stabilization of p27. Purified quiescent transcriptionally activated by p53. Similarly, the host gene B16 mouse melanoma cells expressed higher levels of of miRNA-455-3p, collagen alpha-1 (XXVII) chain, was also a miRNA-27b-3p and miRNA-455-3p relative to their prolif- p53 transcriptional target. Collectively, our results identify erating counterparts. Induction of quiescence resulted in miRNA-27b-3p and miRNA-455-3p as important regulators increased levels of these miRNAs in diverse types of human of cancer cell quiescence in response to p53 and suggest that cancer cell lines. Inhibition of miRNA-27b-3p or miRNA- manipulating miRNA-27b-3p and miRNA-455-3p may con- 455-3p reduced, whereas its overexpression increased, the stitute novel therapeutic avenues for improving outcomes of proportion of quiescent cells in the population, indicating cancer treatment.
    [Show full text]
  • Porcine Circovirus 2 Induction of ROS Is Responsible for Mitophagy in PK-15 Cells Via Activation of Drp1 Phosphorylation
    viruses Article Porcine Circovirus 2 Induction of ROS Is Responsible for Mitophagy in PK-15 Cells via Activation of Drp1 Phosphorylation Yikai Zhang, Renjie Sun, Xiaoliang Li * and Weihuan Fang * Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China; [email protected] (Y.Z.); [email protected] (R.S.) * Correspondence: [email protected] (X.L.); [email protected] (W.F.); Tel.: +86-571-88982291 (X.L.); +86-571-88982242 (W.F.) Received: 2 February 2020; Accepted: 4 March 2020; Published: 6 March 2020 Abstract: Mitochondrial dynamics is essential for the maintenance of cell homeostasis. Previous studies have shown that porcine circovirus 2 (PCV2) infection decreases the mitochondrial membrane potential and causes the elevation of reactive oxygen species (ROS), which may ultimately lead to mitochondrial apoptosis. However, whether PCV2 induce mitophagy remains unknown. Here we show that PCV2-induced mitophagy in PK-15 cells via Drp1 phosphorylation and PINK1/Parkin activation. PCV2 infection enhanced the phosphorylation of Drp1 and its subsequent translocation to mitochondria. PCV2-induced Drp1 phosphorylation could be suppressed by specific CDK1 inhibitor RO-3306, suggesting CDK1 as its possible upstream molecule. PCV2 infection increased the amount of ROS, up-regulated PINK1 expression, and stimulated recruitment of Parkin to mitochondria. N-acetyl-L-cysteine (NAC) markedly decreased PCV2-induced ROS, down-regulated Drp1 phosphorylation, and lessened PINK1 expression and mitochondrial accumulation of Parkin. Inhibition of Drp1 by mitochondrial division inhibitor-1 Mdivi-1 or RNA silencing not only resulted in the reduction of ROS and PINK1, improved mitochondrial mass and mitochondrial membrane potential, and decreased mitochondrial translocation of Parkin, but also led to reduced apoptotic responses.
    [Show full text]
  • The Cyclin-Dependent Kinase Inhibitor and Tumor Suppressor Locus P16/INK4A- P14arf and Regulation of the Transition Into and out of the Cell Cycle
    The Cyclin-Dependent Kinase Inhibitor and Tumor Suppressor Locus p16/INK4A- p14ARF and Regulation of the Transition Into and Out of the Cell Cycle by Payal Agarwal A dissertation submitted to the Graduate Faculty of Auburn University in partial fulfillment of the requirements for the Degree of Doctor of Philosophy Auburn, Alabama May 7, 2012 Copyright, 2011 by Payal Agarwal Approved by Richard C. Bird, Chair, Professor of Pathobiology Bruce F. Smith, Professor of Pathobiology Frederik W. van Ginkel, Associate Professor of Pathobiology Anthony G. Moss, Associate Professor of Biological Sciences Abstract p16/INK4A/CDKN2A is an important tumor suppressor gene located in the INK4A/ARF locus, which encodes a 16 kDa protein known as p16, and a 14 kDa protein known as p14ARF in humans. p16 arrests cell cycle in early G1 phase thereby inhibiting the binding of cyclin dependent kinase 4/6 with cyclinD1. This leaves the retinoblastoma protein (pRb) tumor suppressor hypo-phosphorylated and S phase transcription factor E2F bound and inactive. p14ARF expression up-regulates cyclin dependent kinase inhibitor p21, which inhibits the G1/S phase transition by stabilizing p53 expression upon disassociation from mdm2. We hypothesized that p16 has a role in exit from the cell cycle, becomes defective in cancer cells and has binding partners other than CDK4/CDK6 in quiescent or differentiated cells when their canonical target proteins are thought to be nonfunctional. We have hypothesized that INK4A/ARF encoded proteins perform important regulatory roles that are defective in canine mammary cancer and may cause loss of differentiation potential. Well characterized p16-defective canine mammary cancer cell lines, normal canine fibroblasts, and CMT-derived p16-transfected CMT cell clones, are used to investigate expression of p16 after serum starvation into quiescence followed by re-feeding to induce cell cycle re-entry.
    [Show full text]
  • Exit from Quiescence Displays a Memory of Cell Growth and Division
    ARTICLE DOI: 10.1038/s41467-017-00367-0 OPEN Exit from quiescence displays a memory of cell growth and division Xia Wang1,2, Kotaro Fujimaki1, Geoffrey C. Mitchell1,3, Jungeun Sarah Kwon1, Kimiko Della Croce1, Chris Langsdorf4, Hao Helen Zhang5 & Guang Yao1,6 Reactivating quiescent cells to proliferate is critical to tissue repair and homoeostasis. Quiescence exit is highly noisy even for genetically identical cells under the same environmental conditions. Deregulation of quiescence exit is associated with many diseases, but cellular mechanisms underlying the noisy process of exiting quiescence are poorly understood. Here we show that the heterogeneity of quiescence exit reflects a memory of preceding cell growth at quiescence induction and immediate division history before quiescence entry, and that such a memory is reflected in cell size at a coarse scale. The deterministic memory effects of preceding cell cycle, coupled with the stochastic dynamics of an Rb-E2F bistable switch, jointly and quantitatively explain quiescence-exit heterogeneity. As such, quiescence can be defined as a distinct state outside of the cell cycle while displaying a sequential cell order reflecting preceding cell growth and division variations. 1 Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA. 2 School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, China. 3 Department of Biology, Wofford College, Spartanburg, SC 29303, USA. 4 Molecular Probes, Thermo Fisher Scientific, Eugene, OR 97402, USA. 5 Department of Mathematics, University of Arizona, Tucson, AZ 85721, USA. 6 Arizona Cancer Centre, University of Arizona, Tucson, AZ 85719, USA. Xia Wang, Kotaro Fujimaki and Geoffrey C.
    [Show full text]
  • (PCV2) in Intestinal Porcine Epithelial Cell Line (IPEC-J2) and Interaction Between PCV2 and IPEC-J2 Microfilaments Mengfei Yan, Liqi Zhu and Qian Yang*
    Yan et al. Virology Journal 2014, 11:193 http://www.virologyj.com/content/11/1/193 RESEARCH Open Access Infection of Porcine Circovirus 2 (PCV2) in Intestinal Porcine Epithelial Cell Line (IPEC-J2) and Interaction between PCV2 and IPEC-J2 Microfilaments Mengfei Yan, Liqi Zhu and Qian Yang* Abstract Background: Porcine circovirus-associated disease (PCVAD) is caused by a small pathogenic DNA virus, Porcine circovirus type 2 (PCV2), and is responsible for severe economic losses. PCV2-associated enteritis appears to be a distinct clinical manifestation of PCV2. Most studies of swine enteritis have been performed in animal infection models, but none have been conducted in vitro using cell lines of porcine intestinal origin. An in vitro system would be particularly useful for investigating microfilaments, which are likely to be involved in every stage of the viral lifecycle. Methods: We confirmed that PCV2 infects the intestinal porcine epithelial cell line IPEC-J2 by means of indirect immunofluorescence, transmission electron microscopy, flow cytometry and qRT-PCR. PCV2 influence on microfilaments in IPEC-J2 cells was detected by fluorescence microscopy and flow cytometry. We used Cytochalasin D or Cucurbitacin E to reorganize microfilaments, and observed changes in PCV2 invasion, replication and release in IPEC-J2 cells by qRT-PCR. Results: PCV2 infection changes the ultrastructure of IPEC-J2 cells. PCV2 copy number in IPEC-J2 cells shows a rising trend as infection proceeds. Microfilaments are polymerized at 1 h p.i., but densely packed actin stress fibres are disrupted and total F-actin increases at 24, 48 and 72 h p.i. After Cytochalasin D treatment, invasion of PCV2 is suppressed, while invasion is facilitated by Cucurbitacin E.
    [Show full text]
  • Rotavirus Vaccine Information Statement
    VACCINE INFORMATION STATEMENT Many Vaccine Information Statements are available in Spanish and other languages. Rotavirus Vaccine: See www.immunize.org/vis Hojas de información sobre vacunas están disponibles en español y en muchos otros What You Need to Know idiomas. Visite www.immunize.org/vis Has severe combined immunodeficiency (SCID). Why get vaccinated? 1 Has had a type of bowel blockage called intussusception. Rotavirus vaccine can prevent rotavirus disease. In some cases, your child’s health care provider may Rotavirus causes diarrhea, mostly in babies and decide to postpone rotavirus vaccination to a future young children. The diarrhea can be severe, and lead visit. to dehydration. Vomiting and fever are also common in babies with rotavirus. Infants with minor illnesses, such as a cold, may be vaccinated. Infants who are moderately or severely ill should usually wait until they recover before getting 2 Rotavirus vaccine rotavirus vaccine. Rotavirus vaccine is administered by putting drops Your child’s health care provider can give you more in the child’s mouth. Babies should get 2 or 3 doses information. of rotavirus vaccine, depending on the brand of vaccine used. Risks of a vaccine reaction The first dose must be administered before 15 4 weeks of age. Irritability or mild, temporary diarrhea or vomiting The last dose must be administered by 8 months can happen after rotavirus vaccine. of age. Intussusception is a type of bowel blockage that is Almost all babies who get rotavirus vaccine will be treated in a hospital and could require surgery. It protected from severe rotavirus diarrhea. happens naturally in some infants every year in the Another virus called porcine circovirus (or parts United States, and usually there is no known reason of it) can be found in rotavirus vaccine.
    [Show full text]